101
|
Debédat J, Le Roy T, Voland L, Belda E, Alili R, Adriouch S, Bel Lassen P, Kasahara K, Hutchison E, Genser L, Torres L, Gamblin C, Rouault C, Zucker JD, Kapel N, Poitou C, Marcelin G, Rey FE, Aron-Wisnewsky J, Clément K. The human gut microbiota contributes to type-2 diabetes non-resolution 5-years after Roux-en-Y gastric bypass. Gut Microbes 2022; 14:2050635. [PMID: 35435140 PMCID: PMC9037437 DOI: 10.1080/19490976.2022.2050635] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is efficient at inducing drastic albeit variable weight loss and type-2 diabetes (T2D) improvements in patients with severe obesity and T2D. We hypothesized a causal implication of the gut microbiota (GM) in these metabolic benefits, as RYGB is known to deeply impact its composition. In a cohort of 100 patients with baseline T2D who underwent RYGB and were followed for 5-years, we used a hierarchical clustering approach to stratify subjects based on the severity of their T2D (Severe vs Mild) throughout the follow-up. We identified via nanopore-based GM sequencing that the more severe cases of unresolved T2D were associated with a major increase of the class Bacteroidia, including 12 species comprising Phocaeicola dorei, Bacteroides fragilis, and Bacteroides caecimuris. A key observation is that patients who underwent major metabolic improvements do not harbor this enrichment in Bacteroidia, as those who presented mild cases of T2D at all times. In a separate group of 36 patients with similar baseline clinical characteristics and preoperative GM sequencing, we showed that this increase in Bacteroidia was already present at baseline in the most severe cases of T2D. To explore the causal relationship linking this enrichment in Bacteroidia and metabolic alterations, we selected 13 patients across T2D severity clusters at 5-years and performed fecal matter transplants in mice. Our results show that 14 weeks after the transplantations, mice colonized with the GM of Severe donors have impaired glucose tolerance and insulin sensitivity as compared to Mild-recipients, all in the absence of any difference in body weight and composition. GM sequencing of the recipient animals revealed that the hallmark T2D-severity associated bacterial features were transferred and were associated with the animals' metabolic alterations. Therefore, our results further establish the GM as a key contributor to long-term glucose metabolism improvements (or lack thereof) after RYGB.
Collapse
Affiliation(s)
- Jean Debédat
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Tiphaine Le Roy
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Lise Voland
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | | | - Rohia Alili
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Solia Adriouch
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Pierre Bel Lassen
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Kazuyuki Kasahara
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Evan Hutchison
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Laurent Genser
- Visceral Surgery Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Licia Torres
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Camille Gamblin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Christine Rouault
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Jean-Daniel Zucker
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Universités, Institut de Recherche pour le Développement (IRD), France
| | - Nathalie Kapel
- Functional Coprology Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, France
| | - Christine Poitou
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France
| | - Geneviève Marcelin
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Judith Aron-Wisnewsky
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,CONTACT Judith Aron-Wisnewsky Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Nutrition and obesities; systemic approaches (NutriOmics)75013, Paris, France
| | - Karine Clément
- Nutrition and obesities; systemic approaches (NutriOmics), Sorbonne Université, INSERM, ParisFrance,Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, France,Karine Clément Nutrition and obesities; systemic approaches (NutriOmics) Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, ParisFranceNutrition Department
| |
Collapse
|
102
|
Guo Y, Liu C, Zhao X, Zhang X, Wu Q, Wang Z, Lu J. Changes in gut microbiota, metabolite SCFAs, and GPR43 expression in obese diabetic mice after sleeve gastrectomy. J Appl Microbiol 2022; 133:555-568. [PMID: 35437874 DOI: 10.1111/jam.15583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022]
Abstract
AIMS To evaluate changes in short-chain fatty acid levels and G protein-coupled receptor 43 expression and distribution in gut microbiota and explore their relationships in obese diabetic mice after sleeve gastrectomy. METHODS AND RESULTS Diet-induced obese mice and obese diabetic ob/ob mice were established. Changes in glucose metabolism, lipid metabolism, gut microbiota, metabolite short-chain fatty acids, and G protein-coupled receptor 43 expression were assessed in both models 10 weeks postoperatively. Mice that underwent sleeve gastrectomy exhibited sustained weight loss and reduced glucose, insulin, leptin, and cholesterol levels. Metagenomic sequencing revealed significant characteristic alterations in gut microbiota after sleeve gastrectomy, which were correlated with changes in fecal short-chain fatty acid levels. Postoperatively, G protein-coupled receptor 43 expression in the colon tissue was upregulated in both models, whereas its expression in the adipose tissue was downregulated in the diet-induced obese mouse model. CONCLUSIONS Metabolic improvement in obese and diabetic mice after sleeve gastrectomy is associated with alterations in gut microbiota, short-chain fatty acid levels, and G protein-coupled receptor 43 expression. SIGNIFICANCE AND IMPACT OF STUDY Our findings reveal a possible mechanism through which sleeve gastrectomy improves obesity and diabetes via changes in bacteria producing short-chain fatty acids and G protein-coupled receptor 43.
Collapse
Affiliation(s)
- Yan Guo
- Department of Endocrinology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Chaoqian Liu
- Department of General surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Xiang Zhao
- Department of General surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Xianfang Zhang
- Department of General surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Qingzhi Wu
- Department of General surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Zhijie Wang
- Department of General surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Jin Lu
- Department of Endocrinology, Changhai Hospital, the Second Military Medical University, Shanghai, China
| |
Collapse
|
103
|
Wise J, Plescia T, Cummings BP, Lyo V. Exploring the Relationship Between Bariatric Surgery and Inflammatory Bowel Disease: A Systematic Review. CROHN'S & COLITIS 360 2022; 4:otac013. [PMID: 36777046 PMCID: PMC9802289 DOI: 10.1093/crocol/otac013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Indexed: 11/14/2022] Open
Abstract
Background Obesity affects over 40% of Americans. Bariatric surgery is an increasingly popular and well-studied method to achieve weight loss, improve metabolic homeostasis, and resolve obesity-related comorbid conditions. While the impact of bariatric surgery on weight loss and metabolic health has been extensively studied, there is an increasing body of literature characterizing the impact of bariatric surgery on gastrointestinal health and inflammation. Inflammatory bowel disease (IBD) leads to inflammation in both the small and large intestine, and leads to significant patient morbidity. Similar to obesity, the incidence of IBD is also rising. Patients with IBD and obesity may seek bariatric surgery. The impact of bariatric surgery on IBD is not well understood, but critical to understand for optimal patient care. Herein, we review the currently available literature on the impact of bariatric surgery on IBD including common trends, discrepancies in findings, and remaining knowledge gaps in need of further study. Methods A systematic review of the PubMed/MEDLINE database using PRISMA guidelines was performed. Results We identified 12 manuscripts discussing de novo IBD after bariatric surgery and 16 studying bariatric surgery in patients with pre-existing IBD. Overall, bariatric surgery appears to be safe in patients with pre-existing IBD but may increase the risk of developing de novo IBD. Conclusions Further research into optimal surgical approaches, patient selection, and mechanisms on how bariatric surgery impacts IBD is needed.
Collapse
Affiliation(s)
- Journey Wise
- Department of Biomedical Sciences, Cornell University, School of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Trevor Plescia
- Department of Surgery, University of California, Davis, Sacramento, CA 95817, USA
| | | | - Victoria Lyo
- Department of Surgery, University of California, Davis, Sacramento, CA 95817, USA,Address correspondence to: Victoria Lyo, MD, MTM, UC Davis Medical Center, 2335 Stockton Blvd., NAOB 6113, Sacramento, CA 95817, USA ()
| |
Collapse
|
104
|
Catalán V, Avilés-Olmos I, Rodríguez A, Becerril S, Fernández-Formoso JA, Kiortsis D, Portincasa P, Gómez-Ambrosi J, Frühbeck G. Time to Consider the "Exposome Hypothesis" in the Development of the Obesity Pandemic. Nutrients 2022; 14:1597. [PMID: 35458158 PMCID: PMC9032727 DOI: 10.3390/nu14081597] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
The obesity epidemic shows no signs of abatement. Genetics and overnutrition together with a dramatic decline in physical activity are the alleged main causes for this pandemic. While they undoubtedly represent the main contributors to the obesity problem, they are not able to fully explain all cases and current trends. In this context, a body of knowledge related to exposure to as yet underappreciated obesogenic factors, which can be referred to as the "exposome", merits detailed analysis. Contrarily to the genome, the "exposome" is subject to a great dynamism and variability, which unfolds throughout the individual's lifetime. The development of precise ways of capturing the full exposure spectrum of a person is extraordinarily demanding. Data derived from epidemiological studies linking excess weight with elevated ambient temperatures, in utero, and intergenerational effects as well as epigenetics, microorganisms, microbiota, sleep curtailment, and endocrine disruptors, among others, suggests the possibility that they may work alone or synergistically as several alternative putative contributors to this global epidemic. This narrative review reports the available evidence on as yet underappreciated drivers of the obesity epidemic. Broadly based interventions are needed to better identify these drivers at the same time as stimulating reflection on the potential relevance of the "exposome" in the development and perpetuation of the obesity epidemic.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Iciar Avilés-Olmos
- Department of Neurology, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | | | - Dimitrios Kiortsis
- Department of Nuclear Medicine, Medical School, University of Ioannina, 45110 Ioannina, Greece;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.C.); (A.R.); (S.B.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31008 Pamplona, Spain;
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
105
|
Martinou E, Stefanova I, Iosif E, Angelidi AM. Neurohormonal Changes in the Gut-Brain Axis and Underlying Neuroendocrine Mechanisms following Bariatric Surgery. Int J Mol Sci 2022; 23:3339. [PMID: 35328759 PMCID: PMC8954280 DOI: 10.3390/ijms23063339] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity is a complex, multifactorial disease that is a major public health issue worldwide. Currently approved anti-obesity medications and lifestyle interventions lack the efficacy and durability needed to combat obesity, especially in individuals with more severe forms or coexisting metabolic disorders, such as poorly controlled type 2 diabetes. Bariatric surgery is considered an effective therapeutic modality with sustained weight loss and metabolic benefits. Numerous genetic and environmental factors have been associated with the pathogenesis of obesity, while cumulative evidence has highlighted the gut-brain axis as a complex bidirectional communication axis that plays a crucial role in energy homeostasis. This has led to increased research on the roles of neuroendocrine signaling pathways and various gastrointestinal peptides as key mediators of the beneficial effects following weight-loss surgery. The accumulate evidence suggests that the development of gut-peptide-based agents can mimic the effects of bariatric surgery and thus is a highly promising treatment strategy that could be explored in future research. This article aims to elucidate the potential underlying neuroendocrine mechanisms of the gut-brain axis and comprehensively review the observed changes of gut hormones associated with bariatric surgery. Moreover, the emerging role of post-bariatric gut microbiota modulation is briefly discussed.
Collapse
Affiliation(s)
- Eirini Martinou
- Department of Upper Gastrointestinal Surgery, Frimley Health NHS Foundation Trust, Camberley GU16 7UJ, UK;
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Irena Stefanova
- Department of General Surgery, Frimley Health NHS Foundation Trust, Camberley GU16 7UJ, UK;
| | - Evangelia Iosif
- Department of General Surgery, Royal Surrey County Hospital, Guildford GU2 7XX, UK;
| | - Angeliki M. Angelidi
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
106
|
Camastra S, Palumbo M, Santini F. Nutrients handling after bariatric surgery, the role of gastrointestinal adaptation. Eat Weight Disord 2022; 27:449-461. [PMID: 33895917 PMCID: PMC8933374 DOI: 10.1007/s40519-021-01194-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/10/2021] [Indexed: 01/19/2023] Open
Abstract
Bariatric surgery determines a rearrangement of the gastrointestinal tract that influences nutrient handling and plays a role in the metabolic changes observed after surgery. Most of the changes depend on the accelerated gastric emptying observed in Roux-en-Y gastric bypass (RYGB) and, to a lesser extent, in sleeve gastrectomy (SG). The rapid delivery of meal into the jejunum, particularly after RYGB, contributes to the prompt appearance of glucose in peripheral circulation. Glucose increase is the principal determinant of GLP-1 increase with the consequent stimulation of insulin secretion, the latter balanced by a paradoxical glucagon increase that stimulates EGP to prevent hypoglycaemia. Protein digestion and amino acid absorption appear accelerated after RYGB but not after SG. After RYGB, the adaptation of the gut to the new condition participates to the metabolic change. The intestinal transit is delayed, the gut microbioma is changed, the epithelium becomes hypertrophic and increases the expression of glucose transporter and of the number of cell secreting hormones. These changes are not observed after SG. After RYGB-less after SG-bile acids (BA) increase, influencing glucose metabolism probably modulating FXR and TGR5 with an effect on insulin sensitivity. Muscle, hepatic and adipose tissue insulin sensitivity improve, and the gut reinforces the recovery of IS by enhancing glucose uptake and through the effect of the BA. The intestinal changes observed after RYGB result in a light malabsorption of lipid but not of carbohydrate and protein. In conclusion, functional and morphological adaptations of the gut after RYGB and SG activate inter-organs cross-talk that modulates the metabolic changes observed after surgery.Level of evidence Level V, narrative literature review.
Collapse
Affiliation(s)
- Stefania Camastra
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy. .,Interdepartmental Research Center "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.
| | - Maria Palumbo
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy
| | - Ferruccio Santini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy.,Interdepartmental Research Center "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| |
Collapse
|
107
|
Shao Y, Evers SS, Shin JH, Ramakrishnan SK, Bozadjieva-Kramer N, Yao Q, Shah YM, Sandoval DA, Seeley RJ. Vertical sleeve gastrectomy increases duodenal Lactobacillus spp. richness associated with the activation of intestinal HIF2α signaling and metabolic benefits. Mol Metab 2022; 57:101432. [PMID: 34998940 PMCID: PMC8790500 DOI: 10.1016/j.molmet.2022.101432] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/25/2021] [Accepted: 01/01/2022] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Vertical Sleeve Gastrectomy (VSG) is one of the most efficacious treatments for obesity and its comorbidities. Although a range of evidence suggests that alterations of the microbiota in the distal gut following VSG are pivotal to these metabolic improvements, the effect of surgery to alter the microbiota of the proximal intestine and its effect on host physiology remain largely unknown. As the main bacteria in the upper small intestine, Lactobacillus subspecies have been appreciated as important regulators of gut function. These bacteria also regulate intestinal Hypoxia- Inducible Factor 2α (HIF2α) signaling that plays an integral role in gut physiology and iron absorption. In the present study, we sought to determine the impact of VSG on Lactobacillus spp. in the small intestine and potential downstream impacts of Lactobacillus spp. on HIF2α, specifically in the duodenum. METHODS To determine the effects of VSG on the microbiota and HIF2α signaling in the duodenum, VSG surgeries were performed on diet-induced obese mice. To further probe the relationship between Lactobacillus spp. and HIF2α signaling in the duodenum, we applied a customized high-fat but iron-deficient diet on mice to increase duodenal HIF2α signaling and determined alterations of gut bacteria. To explore the causal role of Lactobacillus spp. in duodenal HIF2α signaling activation, we chronically administered probiotics containing Lactobacillus spp. to high-fat-fed obese mice. Lastly, we studied the effect of lactate, the major metabolite of Lactobacilli, on HIF2α in ex vivo duodenal organoids. RESULTS There were pronounced increases in the abundance of Lactobacillus spp. in samples isolated from duodenal epithelium in VSG-operated mice as compared to sham-operated mice. This was accompanied by an increase in the expression of genes that are targets of HIF2α in the duodenum of VSG-treated mice. Activating HIF2α signaling with a high-fat but iron-deficient diet resulted in weight loss, improvements in glucose regulation, and increased Lactobacillus spp. richness in the duodenum as compared to mice on an iron-replete diet. Chronic administration of probiotics containing Lactobacillus spp. not only increased HIF2α signaling in the duodenum such as occurs after VSG but also resulted in reduced weight gain and improved glucose tolerance in high-fat-fed mice. Furthermore, lactate was able to activate HIF2α in ex vivo duodenal organoids. CONCLUSIONS These results support a model whereby VSG increases duodenal Lactobacillus richness and potentially stimulates intestinal HIF2α signaling via increased lactate production.
Collapse
Affiliation(s)
- Yikai Shao
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center for Obesity and Metabolic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Simon S Evers
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Qiyuan Yao
- Center for Obesity and Metabolic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yatrik M Shah
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
108
|
Itoh H, Tanaka M. “Greedy Organs Hypothesis” for sugar and salt in the pathophysiology of non-communicable diseases in relation to sodium-glucose co-transporters in the intestines and the kidney. Metabol Open 2022; 13:100169. [PMID: 35198947 PMCID: PMC8844901 DOI: 10.1016/j.metop.2022.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 11/05/2022] Open
Abstract
Deposition of visceral fat and insulin resistance play central role in the development of non-communicable diseases (NCDs) including obesity, hypertension and type 2 diabetes. However, we shed more light upon the intestines and the kidney as a strong driver of NCDs. Based upon unexpected outcomes of clinical trials using sodium-glucose cotransporter (SGLT) 2 inhibitors to demonstrate their actions for not only body weight reduction and blood glucose fall but also remarkable cardiorenal protection, we speculate that hyperfunction of the intestines and the kidney is one of critical contributing factors for initiation of NCDs. By detecting high amount of glucose and sodium chloride around them by sweet/salt taste sensors, the intestines and the kidney are designed to (re)absorb these nutrients by up-regulating SGLT1 or SGLT2. We designate these hyperfunctioning organs for nutrient uptake as “greedy organs”. The greedy organs can induce NCDs (“greedy organ hypothesis”). SGLTs are regulated by glucose and sodium chloride, and SGLTs or other genes can be “greedy genes.” Regulating factors for greedy organs are renin-angiotensin system, renal sympathetic nervous activity, gut inflammation/microbiota or oxidative stress. Mitigation of organ greediness by SGLT2 inhibitors, ketone bodies, bariatric surgery, and regular lifestyle to keep rhythmicity of biological clock are promising. We propose the concept of “Greedy Organs” hypothesis as a possible cause of NCDs. Clinical implication of greedy kidney is supported by the effect of SGLT2 inhibitors. The significance of greedy intestines is suggested by the effect of bariatric surgery. The intestines and kidney become hyperactive through upregulation of SGLT1 or 2. To mitigate “greedy organs” should be a promising strategy against NCDs.
Collapse
|
109
|
Martínez-Montoro JI, Kuchay MS, Balaguer-Román A, Martínez-Sánchez MA, Frutos MD, Fernández-García JC, Ramos-Molina B. Gut microbiota and related metabolites in the pathogenesis of nonalcoholic steatohepatitis and its resolution after bariatric surgery. Obes Rev 2022; 23:e13367. [PMID: 34729904 DOI: 10.1111/obr.13367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in parallel with the rising prevalence of obesity, leading to major health and socioeconomic consequences. To date, the most effective therapeutic approach for NAFLD is weight loss. Accordingly, bariatric surgery (BS), which produces marked reductions in body weight, is associated with significant histopathological improvements in advanced stages of NAFLD, such as nonalcoholic steatohepatitis (NASH) and liver fibrosis. BS is also associated with substantial taxonomical and functional alterations in gut microbiota, which are believed to play a significant role in metabolic improvement after BS. Interestingly, gut microbiota and related metabolites may be implicated in the pathogenesis of NAFLD through diverse mechanisms, including specific microbiome signatures, short chain fatty acid production or the modulation of one-carbon metabolism. Moreover, emerging evidence highlights the potential association between gut microbiota changes after BS and NASH resolution. In this review, we summarize the current knowledge on the relationship between NAFLD severity and gut microbiota, as well as the role of the gut microbiome and related metabolites in NAFLD improvement after BS.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Malaga (IBIMA), Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mohammad Shafi Kuchay
- Division of Endocrinology and Diabetes, Medanta - The Medicity Hospital, Gurugram, Haryana, India
| | - Andrés Balaguer-Román
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain.,Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | | | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - José Carlos Fernández-García
- Department of Endocrinology and Nutrition, Regional University Hospital of Malaga, Institute of Biomedical Research in Malaga (IBIMA), Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Bruno Ramos-Molina
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
110
|
Mukorako P, Lemoine N, Biertho L, Lebel S, Roy MC, Plamondon J, Tchernof A, Varin TV, Anhê FF, St-Pierre DH, Marette A, Richard D. Consistent gut bacterial and short-chain fatty acid signatures in hypoabsorptive bariatric surgeries correlate with metabolic benefits in rats. Int J Obes (Lond) 2022; 46:297-306. [PMID: 34686781 DOI: 10.1038/s41366-021-00973-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 07/08/2021] [Accepted: 09/16/2021] [Indexed: 01/16/2023]
Abstract
OBJECTIVE The study aimed at comparing how changes in the gut microbiota are associated to the beneficial effects of the most clinically efficient hypoabsorptive bariatric procedures, namely Roux-en-Y gastric bypass (RYGB), biliopancreatic diversion with duodenal switch (BPD-DS) and single anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI-S). METHODS Diet-induced obese (DIO) male Wistar rats were divided into seven groups. In addition to the groups subjected to RYGB, BPD-DS and SADI-S, the following four control groups were included: SHAM-operated rats fed a high-fat diet (SHAM HF), SHAM fed a low-fat diet (SHAM LF), SHAM HF-pair-weighed to BPD-DS (SHAM HF-PW) and sleeve-gastrectomy (SG) rats. Body weight, food intake, glucose tolerance, insulin sensitivity/resistance, and L-cell secretion were assessed. The gut microbiota (16 S ribosomal RNA gene sequencing) as well as the fecal and cæcal contents of short-chain fatty acids (SCFAs) were also analyzed prior to, and after the surgeries. RESULTS The present study demonstrates the beneficial effect of RYGB, BPD-DS and SADI-S on fat mass gain and glucose metabolism in DIO rats. These benefits were proportional to the effect of the surgeries on food digestibility (BPD-DS > SADI-S > RYGB). Notably, hypoabsorptive surgeries led to consonant microbial signatures characterized by decreased abundance of the Ruminococcaceae (Oscillospira and Ruminococcus), Oscillospiraceae (Oscillibacter) and Christensenellaceae, and increased abundance of the Clostridiaceae (Clostridium), Sutterellaceae (Sutterella) and Enterobacteriaceae. The gut bacteria following hypoabsorptive surgeries were associated with higher fecal levels of propionate, butyrate, isobutyrate and isovalerate. Increases in the fecal SCFAs were in turn positively and strongly correlated with the levels of peptide tyrosine-tyrosine (PYY) and with the beneficial effects of the surgery. CONCLUSION The present study emphasizes the consistency with which the three major hypoabsorptive bariatric procedures RYGB, BPD-DS and SADI-S create a gut microbial environment capable of producing a SCFA profile favorable to the secretion of PYY and to beneficial metabolic effects.
Collapse
Affiliation(s)
- Paulette Mukorako
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.,Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Natacha Lemoine
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Laurent Biertho
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.,Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Stéfane Lebel
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.,Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Marie-Claude Roy
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - Julie Plamondon
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | - André Tchernof
- Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada
| | | | - Fernando F Anhê
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute and Center for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - David H St-Pierre
- Institute of Nutrition and Functional Foods, Québec, QC, Canada.,Department of Exercise Sciences, Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.,Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada.,Institute of Nutrition and Functional Foods, Québec, QC, Canada
| | - Denis Richard
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada. .,Québec Heart and Lung Institute, Chemin Sainte-Foy, Québec, QC, Canada.
| |
Collapse
|
111
|
Abstract
Currently, there are no approved medications to treat patients with nonalcoholic steatohepatitis (NASH) with fibrosis or cirrhosis. Although the management goal in these patients is weight reduction by 7-10% with lifestyle modifications, only less than 10% of patients achieve this target at 1-year, and fewer maintain the weight loss at 5 years. Bariatric surgery is an option that may be considered in those who fail to lose weight by lifestyle changes. Bariatric surgery has been shown to improve liver histology including fibrosis secondary to NASH, in addition to other benefits including an improvement or resolution of type 2 diabetes mellitus, dyslipidemia, and hypertension, and a reduction of cardiovascular morbidity or mortality. There are no guidelines of bariatric surgery indications for the management of NASH. The purpose of this review is to critically appraise the current knowledge of the role of bariatric surgery and the potential mechanisms for its perceived benefits in the management of patients with NASH-related liver disease.
Collapse
|
112
|
Roux-En-Y Gastric Bypass (RYGB) Surgery during High Liquid Sucrose Diet Leads to Gut Microbiota-Related Systematic Alterations. Int J Mol Sci 2022; 23:ijms23031126. [PMID: 35163046 PMCID: PMC8835548 DOI: 10.3390/ijms23031126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
Roux-en-Y gastric bypass (RYGB) surgery has been proven successful in weight loss and improvement of co-morbidities associated with obesity. Chronic complications such as malabsorption of micronutrients in up to 50% of patients underline the need for additional therapeutic approaches. We investigated systemic RYGB surgery effects in a liquid sucrose diet-induced rat obesity model. After consuming a diet supplemented with high liquid sucrose for eight weeks, rats underwent RYGB or control sham surgery. RYGB, sham pair-fed, and sham ad libitum-fed groups further continued on the diet after recovery. Notable alterations were revealed in microbiota composition, inflammatory markers, feces, liver, and plasma metabolites, as well as in brain neuronal activity post-surgery. Higher fecal 4-aminobutyrate (GABA) correlated with higher Bacteroidota and Enterococcus abundances in RYGB animals, pointing towards the altered enteric nervous system (ENS) and gut signaling. Favorable C-reactive protein (CRP), serine, glycine, and 3-hydroxybutyrate plasma profiles in RYGB rats were suggestive of reverted obesity risk. The impact of liquid sucrose diet and caloric restriction mainly manifested in fatty acid changes in the liver. Our multi-modal approach reveals complex systemic changes after RYGB surgery and points towards potential therapeutic targets in the gut-brain system to mimic the surgery mode of action.
Collapse
|
113
|
Seyfried F, Phetcharaburanin J, Glymenaki M, Nordbeck A, Hankir M, Nicholson JK, Holmes E, Marchesi JR, Li JV. Roux-en-Y gastric bypass surgery in Zucker rats induces bacterial and systemic metabolic changes independent of caloric restriction-induced weight loss. Gut Microbes 2022; 13:1-20. [PMID: 33535876 PMCID: PMC7872092 DOI: 10.1080/19490976.2021.1875108] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mechanisms of Roux-en-Y gastric bypass (RYGB) surgery are not fully understood. This study aimed to investigate weight loss-independent bacterial and metabolic changes, as well as the absorption of bacterial metabolites and bile acids through the hepatic portal system following RYGB surgery. Three groups of obese Zucker (fa/fa) rats were included: RYGB (n = 11), sham surgery and body weight matched with RYGB (Sham-BWM, n = 5), and sham surgery fed ad libitum (Sham-obese, n = 5). Urine and feces were collected at multiple time points, with portal vein and peripheral blood obtained at the end of the study. Metabolic phenotyping approaches and 16S rRNA gene sequencing were used to determine the biochemical and bacterial composition of the samples, respectively. RYGB surgery-induced distinct metabolic and bacterial disturbances, which were independent of weight loss through caloric restriction. RYGB resulted in lower absorption of phenylalanine and choline, and higher urinary concentrations of host-bacterial co-metabolites (e.g., phenylacetylglycine, indoxyl sulfate), together with higher fecal trimethylamine, suggesting enhanced bacterial aromatic amino acid and choline metabolism. Short chain fatty acids (SCFAs) were lower in feces and portal vein blood from RYGB group compared to Sham-BWM, accompanied with lower abundances of Lactobacillaceae, and Ruminococcaceae known to contain SCFA producers, indicating reduced bacterial fiber fermentation. Fecal γ-amino butyric acid (GABA) was found in higher concentrations in RYGB than that in Sham groups and could play a role in the metabolic benefits associated with RYGB surgery. While no significant difference in urinary BA excretion, RYGB lowered both portal vein and circulating BA compared to Sham groups. These findings provide a valuable resource for how dynamic, multi-systems changes impact on overall metabolic health, and may provide potential therapeutic targets for developing downstream non-surgical treatment for metabolic disease.
Collapse
Affiliation(s)
- Florian Seyfried
- Department of General, Visceral, Transplant, Vascular, and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jutarop Phetcharaburanin
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College LondonLondon, UK,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Maria Glymenaki
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College LondonLondon, UK
| | - Arno Nordbeck
- Department of General, Visceral, Transplant, Vascular, and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Mohammed Hankir
- Department of General, Visceral, Transplant, Vascular, and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jeremy K Nicholson
- Division of Organisms and Environment, School of Biosciences, Institute of Health Futures, Murdoch University, Perth, Western Australia, Australia
| | - Elaine Holmes
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College LondonLondon, UK,Division of Organisms and Environment, School of Biosciences, Institute of Health Futures, Murdoch University, Perth, Western Australia, Australia
| | - Julian R. Marchesi
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College LondonLondon, UK,School of Biosciences, Cardiff University, Cardiff, UK
| | - Jia V. Li
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College LondonLondon, UK,CONTACT Jia V. Li Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
114
|
Akalestou E, Miras AD, Rutter GA, le Roux CW. Mechanisms of Weight Loss After Obesity Surgery. Endocr Rev 2022; 43:19-34. [PMID: 34363458 PMCID: PMC8755990 DOI: 10.1210/endrev/bnab022] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 02/07/2023]
Abstract
Obesity surgery remains the most effective treatment for obesity and its complications. Weight loss was initially attributed to decreased energy absorption from the gut but has since been linked to reduced appetitive behavior and potentially increased energy expenditure. Implicated mechanisms associating rearrangement of the gastrointestinal tract with these metabolic outcomes include central appetite control, release of gut peptides, change in microbiota, and bile acids. However, the exact combination and timing of signals remain largely unknown. In this review, we survey recent research investigating these mechanisms, and seek to provide insights on unanswered questions over how weight loss is achieved following bariatric surgery which may eventually lead to safer, nonsurgical weight-loss interventions or combinations of medications with surgery.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alexander D Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore.,University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Carel W le Roux
- Diabetes Complications Research Centre, University College Dublin, Ireland.,Diabetes Research Group, School of Biomedical Science, Ulster University, Belfast, UK
| |
Collapse
|
115
|
Glenny EM, Fouladi F, Thomas SA, Bulik-Sullivan EC, Tang Q, Djukic Z, Trillo-Ordonez YS, Fodor AA, Tarantino LM, M. Bulik C, Carroll IM. Gut microbial communities from patients with anorexia nervosa do not influence body weight in recipient germ-free mice. Gut Microbes 2022; 13:1-15. [PMID: 33769200 PMCID: PMC8007138 DOI: 10.1080/19490976.2021.1897216] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Anorexia nervosa (AN) is a psychiatric disorder that presents with profound weight dysregulation, metabolic disturbances, and an abnormal composition of gut microbial communities. As the intestinal microbiota can influence host metabolism, the impact of enteric microbial communities from patients with AN on host weight and adiposity was investigated. Germ-free (GF) mice were colonized with fecal microbiotas from either patients with AN (n = 4) prior to inpatient treatment (AN T1, n = 50 recipient mice), the same 4 patients following clinical renourishment (AN T2, n = 53 recipient mice), or age- and sex-matched non-AN controls (n = 4 human donors; non-AN, n = 50 recipient mice). Biological and fecal microbiota data were analyzed with linear mixed-effects models. Body weight did not differ significantly between AN recipient mice (T1 and T2) and non-AN recipient mice following 4 weeks of colonization. Enteric microbiotas from recipient mice colonized with AN T1 and AN T2 fecal microbiotas were more similar to each other compared with enteric microbiotas from non-AN recipient mice. Specific bacterial families in the Actinobacteria, Bacteroidetes, and Firmicutes phyla were significantly associated with body weight, fat mass, and cecum weight irrespective of the donor group. These data suggest that body weight, fat mass, and cecum weight of colonized GF mice are associated with human fecal microbes and independent of donor AN status, although additional analyses with larger cohorts are warranted.
Collapse
Affiliation(s)
- Elaine M. Glenny
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,CONTACT Ian M. Carroll Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Farnaz Fouladi
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Stephanie A. Thomas
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily C. Bulik-Sullivan
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Quyen Tang
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Graduate School of Professional Psychology, Morrison Family College of Health, University of St. Thomas, Minneapolis, MN, USA
| | - Zorka Djukic
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yesel S. Trillo-Ordonez
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Lisa M. Tarantino
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cynthia M. Bulik
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
116
|
Tu J, Wang Y, Jin L, Huang W. Bile acids, gut microbiota and metabolic surgery. Front Endocrinol (Lausanne) 2022; 13:929530. [PMID: 36072923 PMCID: PMC9441571 DOI: 10.3389/fendo.2022.929530] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic surgery, or bariatric surgery, is currently the most effective approach for treating obesity and its complications. Vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) are the top two types of commonly performed metabolic surgery now. The precise mechanisms of how the surgeries work are still unclear, therefore much research has been conducted in this area. Gut hormones such as GLP-1 and PYY have been studied extensively in the context of metabolic surgery because they both participate in satiety and glucose homeostasis. Bile acids, whose functions cover intestinal lipid absorption and various aspects of metabolic regulation via the action of FXR, TGR5, and other bile acid receptors, have also been actively investigated as potential mediators of metabolic surgery. Additionally, gut microbiota and their metabolites have also been studied because they can affect metabolic health. The current review summarizes and compares the recent scientific progress made on identifying the mechanisms of RYGB and VSG. One of the long-term goals of metabolic/bariatric surgery research is to develop new pharmacotherapeutic options for the treatment of obesity and diabetes. Because obesity is a growing health concern worldwide, there is a dire need in developing novel non-invasive treatment options.
Collapse
Affiliation(s)
- Jui Tu
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Lihua Jin
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
- *Correspondence: Wendong Huang,
| |
Collapse
|
117
|
Lo T, Lee Y, Tseng CY, Hu Y, Connelly MA, Mantzoros CS, Karp JM, Tavakkoli A. Daily transient coating of the intestine leads to weight loss and improved glucose tolerance. Metabolism 2022; 126:154917. [PMID: 34687727 PMCID: PMC8666968 DOI: 10.1016/j.metabol.2021.154917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Roux-en-Y gastric bypass surgery (RYGB) has been shown to be the gold standard treatment for obesity associated type-2-diabetes (T2D), however many T2D patients do not qualify or are reluctant to proceed with surgery due to its potential risks and permanent changes to GI anatomy. We have previously described a novel oral formulation, LuCI, that provides a transient coating of the proximal bowel and mimics the effects of RYGB. Herein, we aim to investigate the outcome of chronic LuCI administration on weight and glucose homeostasis. METHODS Sprague-Dawley rats on a high fat diet achieving diet-induced obesity (DIO) received 5 weeks of daily LuCI or normal saline as control (n = 8/group). Daily weights and glucose tolerance were monitored throughout the experiment. At 5 weeks, systemic blood was sampled through a surgically placed jugular vein catheter, before and during an intestinal glucose bolus, to investigate changes in key hormones involved in glucose metabolism. To elucidate the effects of LuCI on nutrient absorption, fecal output and food intake were measured simultaneously with the analysis of homogenized stool samples performed using bomb calorimetry. RESULTS At 5 weeks, LuCI animals weighted 8.3% less and had lower fasting glucose levels than Controls (77.6 ± 3.8 mg/dl vs. 99.1 ± 2.7 mg/dl, P < 0.001). LuCI-treated animals had lower baseline insulin and HOMA-IR. Post-prandially, LuCI group had increased GLP-1 and GIP secretion following a glucose challenge. Serum lipid analysis revealed lowered LDL levels highlighting the potential to not only improve glucose control but also modify cardiovascular risk. We then investigated whether LuCI's effect on proximal bowel exclusion may play a role in energy balance. Bomb calorimetry analysis suggested that LuCI reduced calorie absorption with no difference in caloric consumption. CONCLUSION We demonstrated that LuCI recapitulates the physical and hormonal changes seen after RYGB and can ameliorate weight gain and improve insulin sensitivity in a DIO rat model. Since LuCI's effect is transient and without systemic absorption, LuCI has the potential to be a novel therapy for overweight or obese T2D patients.
Collapse
Affiliation(s)
- Tammy Lo
- Laboratory for Surgical and Metabolic Research, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Nanomedicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Boston, MA, USA
| | - Chung-Yi Tseng
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Nanomedicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Boston, MA, USA
| | - Yangshuo Hu
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Nanomedicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Boston, MA, USA
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, NC, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Nanomedicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Boston, MA, USA.
| | - Ali Tavakkoli
- Laboratory for Surgical and Metabolic Research, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of General and GI Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
118
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
119
|
An Z, Wang H, Mokadem M. Role of the Autonomic Nervous System in Mechanism of Energy and Glucose Regulation Post Bariatric Surgery. Front Neurosci 2021; 15:770690. [PMID: 34887725 PMCID: PMC8649921 DOI: 10.3389/fnins.2021.770690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/15/2021] [Indexed: 01/06/2023] Open
Abstract
Even though lifestyle changes are the mainstay approach to address obesity, Sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) are the most effective and durable treatments facing this pandemic and its associated metabolic conditions. The traditional classifications of bariatric surgeries labeled them as “restrictive,” “malabsorptive,” or “mixed” types of procedures depending on the anatomical rearrangement of each one of them. This conventional categorization of bariatric surgeries assumed that the “restrictive” procedures induce their weight loss and metabolic effects by reducing gastric content and therefore having a smaller reservoir. Similarly, the “malabsorptive” procedures were thought to induce their main energy homeostatic effects from fecal calorie loss due to intestinal malabsorption. Observational data from human subjects and several studies from rodent models of bariatric surgery showed that neither of those concepts is completely true, at least in explaining the multiple metabolic changes and the alteration in energy balance that those two surgeries induce. Rather, neuro-hormonal mechanisms have been postulated to underly the physiologic effects of those two most performed bariatric procedures. In this review, we go over the role the autonomic nervous system plays- through its parasympathetic and sympathetic branches- in regulating weight balance and glucose homeostasis after SG and RYGB.
Collapse
Affiliation(s)
- Zhibo An
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Haiying Wang
- Department of Physiology, Basic Medical School of Jining Medical University, Jining, China
| | - Mohamad Mokadem
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, IA, United States.,Obesity Research and Education Initiative, The University of Iowa, Iowa City, IA, United States.,Iowa City Veterans Affairs Health Care System, Iowa City, IA, United States
| |
Collapse
|
120
|
Watanabe M, Sianoya A, Mishima R, Therdtatha P, Rodriguez A, Ramos DC, Lee YK, Dalmacio LM, Nakayama J. Gut microbiome status of urban and rural Filipino adults in relation to diet and metabolic disorders. FEMS Microbiol Lett 2021; 368:6445026. [PMID: 34849762 DOI: 10.1093/femsle/fnab149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Here, we aim to understand the condition of the gut microbiome of Filipino adults in relation to their diet and metabolic status. Compared to rural Albay (n = 67), the gut microbiome of subjects living in urban Manila (n = 25) was more colonized by the order Clostridiales, which was negatively correlated with host carbohydrate consumption. Principal component analysis using the genus composition of the 92 total subjects indicated four microbiome types: one type driven by Prevotella, which was associated with high rice consumption and mainly consisted of healthy Albay subjects, one Clostridiales-driven group containing a number of type 2 diabetes mellitus (T2D) subjects from both Manila and Albay who showed lower butyrate levels in association with a decrease in Mediterraneibacter faecis, and the other two types showing dysbiosis-like microbiomes with Lactobacillus and Bifidobacterium overgrowth, with a high ratio of T2D and obese subjects. Multivariate logistic regression analysis suggested high dietary energy intake, and two Veillonellaeae genera, Dialister and Megasphaera, as T2D risk factors, while Prevotella and M. faecis as anti-T2D factors. In conclusion, low-carbohydrate diets restructured the Prevotella-driven gut microbiome, which may predispose Filipino people with high energy diet to T2D.
Collapse
Affiliation(s)
- Mai Watanabe
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Abraham Sianoya
- Department of Biochemistry and Molecular Biology (DBMB), College of Medicine, University of the Philippines Manila, 547 Pedro Gil Street, Ermita, Manila 1000 Philippines
| | - Riko Mishima
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Phatthanaphong Therdtatha
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Abigail Rodriguez
- Department of Biochemistry and Molecular Biology (DBMB), College of Medicine, University of the Philippines Manila, 547 Pedro Gil Street, Ermita, Manila 1000 Philippines
| | - Donna Christene Ramos
- Department of Biochemistry and Molecular Biology (DBMB), College of Medicine, University of the Philippines Manila, 547 Pedro Gil Street, Ermita, Manila 1000 Philippines
| | - Yuan Kun Lee
- Department of Microbiology and Immunology, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
| | - Leslie Michelle Dalmacio
- Department of Biochemistry and Molecular Biology (DBMB), College of Medicine, University of the Philippines Manila, 547 Pedro Gil Street, Ermita, Manila 1000 Philippines
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
121
|
Zhou H, Tullius SG. Effects of obesity and weight-loss surgery shift the microbiome and impact alloimmune responses. Curr Opin Organ Transplant 2021; 26:603-608. [PMID: 34714789 PMCID: PMC8562884 DOI: 10.1097/mot.0000000000000920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Obesity is a worldwide health problem with increasing rates in both children and adults. Bariatric surgery (BS) represents the only effective long-term treatment. Beneficial effects of BS may be mediated through shifts of the gut microbiome. Here, we introduce data linking the microbiome to alloimmune responses. RECENT FINDINGS The rapid development of microbiome sequencing technologies in addition to the availability of gnotobiotic facilities have enabled mechanistic investigations on modulations of alloimmune responses through microbiomes. BS has been shown to improve comorbidities and chronic inflammation caused by obesity. Changes in microbiota and microbiota-related metabolites may play a role. Patients either listed or having received a transplant have undergone weight loss surgery, thus allowing to dissect mechanisms of microbial shifts to alloimmunity. SUMMARY Weight loss and BS have the potential to improve transplant outcomes by ameliorating alloimmune responses. Those effects may be carried out through alterations of the gut microbiome.
Collapse
Affiliation(s)
- Hao Zhou
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
122
|
Xu E, Chen C, Fu J, Zhu L, Shu J, Jin M, Wang Y, Zong X. Dietary fatty acids in gut health: Absorption, metabolism and function. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:1337-1344. [PMID: 34786506 PMCID: PMC8570925 DOI: 10.1016/j.aninu.2021.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 12/23/2022]
Abstract
In biological responses, fatty acids (FA) are absorbed and metabolized in the form of substrates for energy production. The molecular structures (number of double bonds and chain length) and composition of dietary FA impact digestion, absorption and metabolism, and the biological roles of FA. Recently, increasing evidence indicates that FA are essentially utilized as an energy source and are signaling molecules that exert physiological activity of gut microbiota and immune responses. In addition, FA could serve as natural ligands for orphan G protein-coupled receptors (GPCR), also called free fatty acid receptors (FFAR), which intertwine metabolic and immune systems via multiple mechanisms. The present review explores the recent findings on FA absorption and its impact on gut health, particularly addressing the mechanism by which dietary FA potentially influences intestinal microbiota and epithelial functions. Also, this work attempts to uncover research ideas for devising future strategies for manipulating the composition of dietary FA to regulate gut health and support a normal immune system for metabolic and immune disorders.
Collapse
Affiliation(s)
- E. Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Chao Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Luoyi Zhu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Junlan Shu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| |
Collapse
|
123
|
Yukina MY, Chernova MO, Troshina EA, Evdoshenko VV, Platonova NM. Postprandial hypoglycemia after upper gastrointestinal tract surgery: prevalence and pathophysiology (part 1). ALMANAC OF CLINICAL MEDICINE 2021; 49:285-296. [DOI: 10.18786/2072-0505-2021-49-029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Nowadays obesity is a major health problem worldwide. Surgery is the most promising treatment for morbid obesity. There are two types of bariatric procedures, one to reduce the food intake volume and the other to limit the absorption of nutrients. Despite the indisputable advantages of bariatric surgery for weight loss, it is necessary to remember the potential risk of severe complications, such as hypoglycemic syndrome at 2 to 3 hours after ingestion of rapidly absorbable carbohydrates manifested by adrenergic and neuroglycopenic symptoms. According to the literature, the prevalence of post-bariatric postprandial hyperinsulinemic hypoglycemia (PHH) varies from 10% to 75%. PHH in post-bariatric patients should be differentiated from the syndrome of non-insulinoma pancreatogenic hypoglycemia and from insulinoma; however, these diseases are described also in patients after bariatric surgery.The mechanisms of PHH as an outcome of shunting bariatric procedures are currently not fully clear. According to the recent studies, incretin hypersecretion in response to the accelerated flow of carbohydrates into the small intestine plays a leading role in the inappropriate excess production of insulin by the pancreas. In addition, there are hypotheses on a slower normalization of insulin production during more rapid bodyweight decrease and regression of insulin resistance, the role of alpha-cell dysfunction, disturbed negative feedback between insulin and ghrelin, compensatory hyperplasia and hypertrophy of the remaining enterocytes (including L-cells), changes in gut microflora, bile acids level and composition. A number of other mechanisms have also been proposed that require further studies.
Collapse
Affiliation(s)
| | | | | | - V. V. Evdoshenko
- Institute of Plastic Surgery and Cosmetology; N.I. Pirogov Russian National Research Medical University
| | | |
Collapse
|
124
|
Duggan BM, Tamrakar AK, Barra NG, Anhê FF, Paniccia G, Wallace JG, Stacey HD, Surette MG, Miller MS, Sloboda DM, Schertzer JD. Gut microbiota-based vaccination engages innate immunity to improve blood glucose control in obese mice. Mol Metab 2021; 55:101404. [PMID: 34839023 PMCID: PMC8693341 DOI: 10.1016/j.molmet.2021.101404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
Objective Obesity and diabetes increase circulating levels of microbial components derived from the gut microbiota. Individual bacterial factors (i.e., postbiotics) can have opposing effects on blood glucose. Methods We tested the net effect of gut bacterial extracts on blood glucose in mice using a microbiota-based vaccination strategy. Results Male and female mice had improved glucose and insulin tolerance five weeks after a single subcutaneous injection of a specific dose of a bacterial extract obtained from the luminal contents of the upper small intestine (SI), lower SI, or cecum. Injection of mice with intestinal extracts from germ-free mice revealed that bacteria were required for a microbiota-based vaccination to improve blood glucose control. Vaccination of Nod1−/−, Nod2−/−, and Ripk2−/− mice showed that each of these innate immune proteins was required for bacterial extract injection to improve blood glucose control. A microbiota-based vaccination promoted an immunoglobulin-G (IgG) response directed against bacterial extract antigens, where subcutaneous injection of mice with the luminal contents of the lower SI elicited a bacterial extract-specific IgG response that is compartmentalized to the lower SI of vaccinated mice. A microbiota-based vaccination was associated with an altered microbiota composition in the lower SI and colon of mice. Lean mice only required a single injection of small intestinal-derived bacterial extract, but high fat diet (HFD)-fed, obese mice required prime-boost bacterial extract injections for improvements in blood glucose control. Conclusions Subversion of the gut barrier by vaccination with a microbiota-based extract engages innate immunity to promote long-lasting improvements in blood glucose control in a dose-dependent manner. Subcutaneous injection of gut bacterial extracts improved blood glucose control in mice. Microbiota-based vaccination engaged NOD1-NOD2-RIPK2 to alter blood glucose. Microbiota-based vaccination promoted a proximal gut IgG response. Microbiota-based vaccination altered the composition of the gut microbiome. Obese mice required prime-boost injections to improve blood glucose control.
Collapse
Affiliation(s)
- Brittany M Duggan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Canada
| | - Akhilesh K Tamrakar
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Canada
| | - Fernando F Anhê
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Canada
| | - Gabriella Paniccia
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Jessica G Wallace
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Hannah D Stacey
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada; McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada; Department of Medicine, McMaster University, Hamilton, Canada
| | - Matthew S Miller
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada; McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Department of Obstetrics and Gynecology, McMaster University, Hamilton, Canada; Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Canada.
| |
Collapse
|
125
|
Zhang MW, Fan BS, Yu JG. Case Report: Miles Surgery Ameliorates High Blood Pressure in a Rectal Carcinoma Patient With Essential Hypertension. Front Cardiovasc Med 2021; 8:762959. [PMID: 34796217 PMCID: PMC8593111 DOI: 10.3389/fcvm.2021.762959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Hypertension is one of the major causes of public health problems. Multiple factors affecting gastrointestinal tract function are involved in hypertension. Emerging studies have manifested that gut intervention may play significant roles in regulating blood pressure but the underlying mechanisms are complex and not fully clear. Here, we report a case of 66 years old male who had a long history of hypertension and received Miles surgery for rectal carcinoma. The blood pressure of this patient was returned to normal levels after the operation. The possible reasons could be the modulation of sympathetic tone and the gut microbiota-brain axis. This report provides evidence about the relevance between hypertension and gut intervention particularly in the colorectal sites and gives hints for investigating the possible mechanisms of hypertension and the novel strategy for blood pressure control.
Collapse
Affiliation(s)
- Meng-Wan Zhang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo-Shi Fan
- Department of Thoracic Surgery, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jian-Guang Yu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
126
|
He S, Yu D, Li P, Zhang M, Xing S, Sun C, Li ZH. Triphenyltin exposure causes changes in health-associated gut microbiome and metabolites in marine medaka. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 288:117751. [PMID: 34252717 DOI: 10.1016/j.envpol.2021.117751] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 06/13/2023]
Abstract
Triphenyltin (TPT), an organic compound with a wide range of applications, is often detected in water bodies and aquatic animals. However, the mechanism underlying the biological metabolic health problems caused by long-term exposure to environment concentrations of TPT remains unclear. The morphology and gene expression in the gut and liver were investigated; and 16SrRNA gene amplification sequencing and non-targeted LC-MS/MS metabonomics were investigated after marine medaka (Oryzias melastigma) was treated with 1, 10, and 100 ng/L TPT for 21 days. During prolonged exposure to TPT, the adaptation mechanism maximized the energy of absorption, increased the length of intestinal microvilli, reduced the number of rough endoplasmic reticulum in the liver, and caused loss of weight. TPT exposure significantly changed the intestinal microbiome of marine medaka, thereby resulting in a significant decrease in microbial diversity. Following exposure to 100 ng/L TPT, the metabolic profiles were significantly changed and the altered metabolites were mainly concentrated in the lipid metabolic pathway. Finally, based on comprehensive network analysis, the association between the significantly changed bacteria and metabolites contributed further to the prediction of the impact of TPT on the host. This study provides a novel insight into the underlying mechanisms of host metabolic diseases caused by TPT and emphasizes the importance of monitoring pollutants in the environment.
Collapse
Affiliation(s)
- Shuwen He
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Daode Yu
- Marine Biology Institute of Shandong Province, Qingdao, Shandong, 266104, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Min Zhang
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, Shandong, 266109, China
| | - Shaoying Xing
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Cuici Sun
- State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301, Guangzhou, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| |
Collapse
|
127
|
Blonde GD, Price RK, le Roux CW, Spector AC. Meal Patterns and Food Choices of Female Rats Fed a Cafeteria-Style Diet Are Altered by Gastric Bypass Surgery. Nutrients 2021; 13:3856. [PMID: 34836110 PMCID: PMC8623594 DOI: 10.3390/nu13113856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
After Roux-en-Y gastric bypass surgery (RYGB), rats tend to reduce consumption of high-sugar and/or high-fat foods over time. Here, we sought to investigate the behavioral mechanisms underlying these intake outcomes. Adult female rats were provided a cafeteria diet comprised of five palatable foodstuffs varying in sugar and fat content and intake was monitored continuously. Rats were then assigned to either RYGB, or one of two control (CTL) groups: sham surgery or a nonsurgical control group receiving the same prophylactic iron treatments as RYGB rats. Post-sur-gically, all rats consumed a large first meal of the cafeteria diet. After the first meal, RYGB rats reduced intake primarily by decreasing the meal sizes relative to CTL rats, ate meals more slowly, and displayed altered nycthemeral timing of intake yielding more daytime meals and fewer nighttime meals. Collectively, these meal patterns indicate that despite being motivated to consume a cafeteria diet after RYGB, rats rapidly learn to modify eating behaviors to consume foods more slowly across the entire day. RYGB rats also altered food preferences, but more slowly than the changes in meal patterns, and ate proportionally more energy from complex carbohydrates and protein and proportionally less fat. Overall, the pattern of results suggests that after RYGB rats quickly learn to adjust their size, eating rate, and distribution of meals without altering meal number and to shift their macronutrient intake away from fat; these changes appear to be more related to postingestive events than to a fundamental decline in the palatability of food choices.
Collapse
Affiliation(s)
- Ginger D. Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| | - Ruth K. Price
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK;
| | - Carel W. le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
128
|
Ocaña-Wilhelmi L, Martín-Núñez GM, Ruiz-Limón P, Alcaide J, García-Fuentes E, Gutiérrez-Repiso C, Tinahones FJ, Moreno-Indias I. Gut Microbiota Metabolism of Bile Acids Could Contribute to the Bariatric Surgery Improvements in Extreme Obesity. Metabolites 2021; 11:metabo11110733. [PMID: 34822391 PMCID: PMC8620296 DOI: 10.3390/metabo11110733] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Bariatric surgery is the only procedure to obtain and maintain weight loss in the long term, although the mechanisms driving these benefits are not completely understood. In the last years, gut microbiota has emerged as one of the drivers through its metabolites, especially secondary bile acids. In the current study, we have compared the gut microbiota and the bile acid pool, as well as anthropometric and biochemical parameters, of patient with morbid obesity who underwent bariatric surgery by two different techniques, namely Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG). Gut microbiota populations differed after the respective procedures, particularly with respect to the Enterobacteriaceae family. Both techniques resulted in changes in the bile acids pool, but RYGB was the procedure which suffered the greatest changes, with a reduction in most of their levels. Blautia and Veillonella were the two genera that more relationships showed with secondary bile acids, indicating a possible role in their formation and inhibition, respectively. Correlations with the anthropometric and biochemical variables showed that secondary bile acids could have a role in the amelioration of the glucose and HDL-cholesterol levels. Thus, we have observed a possible relationship between the interaction of the bile acids pool metabolized by the gut microbiota in the metabolic improvements obtained by bariatric surgery in the frame of morbid obesity, deserving further investigation in greater cohorts to decipher the role of each bile acid in the homeostasis of the host for their possible use in the development of microbiota-based therapeutics, such as new drugs, postbiotics or probiotics.
Collapse
Affiliation(s)
- Luis Ocaña-Wilhelmi
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Universidad de Málaga, 29010 Málaga, Spain;
- Unidad de Gestión Clínica de Cirugía General y del Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Gracia María Martín-Núñez
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Patricia Ruiz-Limón
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Juan Alcaide
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Eduardo García-Fuentes
- Department of Gastroenterology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Málaga (IBIMA), Málaga University, 29010 Málaga, Spain;
- CIBER Enfermedades Hepáticas y Digestivas-CIBEREHD, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carolina Gutiérrez-Repiso
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| |
Collapse
|
129
|
Howard EJ, Lam TKT, Duca FA. The Gut Microbiome: Connecting Diet, Glucose Homeostasis, and Disease. Annu Rev Med 2021; 73:469-481. [PMID: 34678047 DOI: 10.1146/annurev-med-042220-012821] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Type 2 diabetes rates continue to rise unabated, underscoring the need to better understand the etiology and potential therapeutic options available for this disease. The gut microbiome plays a role in glucose homeostasis, and diabetes is associated with alterations in the gut microbiome. Given that consumption of a Western diet is associated with increased metabolic disease, and that a Western diet alters the gut microbiome, it is plausible that changes in the gut microbiota mediate the dysregulation in glucose homeostasis. In this review, we highlight a few of the most significant mechanisms by which the gut microbiome can influence glucose regulation, including changes in gut permeability, gut-brain signaling, and production of bacteria-derived metabolites like short-chain fatty acids and bile acids. A better understanding of these pathways could lead to the development of novel therapeutics to target the gut microbiome in order to restore glucose homeostasis in metabolic disease. Expected final online publication date for the Annual Review of Medicine, Volume 73 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Elizabeth J Howard
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona 85721, USA
| | - Tony K T Lam
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada.,Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Frank A Duca
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona 85721, USA.,BIO5 Institute, University of Arizona, Tucson, Arizona 85721, USA;
| |
Collapse
|
130
|
Ben-Haroush Schyr R, Al-Kurd A, Moalem B, Permyakova A, Israeli H, Bardugo A, Arad Y, Hefetz L, Bergel M, Haran A, Azar S, Magenheim I, Tam J, Grinbaum R, Ben-Zvi D. Sleeve Gastrectomy Suppresses Hepatic Glucose Production and Increases Hepatic Insulin Clearance Independent of Weight Loss. Diabetes 2021; 70:2289-2298. [PMID: 34341005 PMCID: PMC8576500 DOI: 10.2337/db21-0251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022]
Abstract
Bariatric operations induce weight loss, which is associated with an improvement in hepatic steatosis and a reduction in hepatic glucose production. It is not clear whether these outcomes are entirely due to weight loss, or whether the new anatomy imposed by the surgery contributes to the improvement in the metabolic function of the liver. We performed vertical sleeve gastrectomy (VSG) on obese mice provided with a high-fat high-sucrose diet and compared them to diet and weight-matched sham-operated mice (WMS). At 40 days after surgery, VSG-operated mice displayed less hepatic steatosis compared with WMS. By measuring the fasting glucose and insulin levels in the blood vessels feeding and draining the liver, we showed directly that hepatic glucose production was suppressed after VSG. Insulin levels were elevated in the portal vein, and hepatic insulin clearance was elevated in VSG-operated mice. The hepatic expression of genes associated with insulin clearance was upregulated. We repeated the experiment in lean mice and observed that portal insulin and glucagon are elevated, but only insulin clearance is increased in VSG-operated mice. In conclusion, direct measurement of glucose and insulin in the blood entering and leaving the liver shows that VSG affects glucose and insulin metabolism through mechanisms independent of weight loss and diet.
Collapse
Affiliation(s)
- Rachel Ben-Haroush Schyr
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abbas Al-Kurd
- Department of Surgery, Hadassah Medical Center-Mt. Scopus, Jerusalem, Israel
| | - Botros Moalem
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Permyakova
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadar Israeli
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aya Bardugo
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yhara Arad
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liron Hefetz
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael Bergel
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Haran
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shahar Azar
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Itia Magenheim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joseph Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronit Grinbaum
- Department of Surgery, Hadassah Medical Center-Mt. Scopus, Jerusalem, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School-The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
131
|
Gut microbiota link dietary fiber intake and short-chain fatty acid metabolism with eating behavior. Transl Psychiatry 2021; 11:500. [PMID: 34599144 PMCID: PMC8486801 DOI: 10.1038/s41398-021-01620-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 02/08/2023] Open
Abstract
The gut microbiome has been speculated to modulate feeding behavior through multiple factors, including short-chain fatty acids (SCFA). Evidence on this relationship in humans is however lacking. We aimed to explore if specific bacterial genera relate to eating behavior, diet, and SCFA in adults. Moreover, we tested whether eating-related microbiota relate to treatment success in patients after Roux-en-Y gastric bypass (RYGB). Anthropometrics, dietary fiber intake, eating behavior, 16S-rRNA-derived microbiota, and fecal and serum SCFA were correlated in young overweight adults (n = 27 (9 F), 21-36 years, BMI 25-31 kg/m2). Correlated genera were compared in RYGB (n = 23 (16 F), 41-70 years, BMI 25-62 kg/m2) and control patients (n = 17 (11 F), 26-69 years, BMI 25-48 kg/m2). In young adults, 7 bacteria genera, i.e., Alistipes, Blautia, Clostridiales cluster XVIII, Gemmiger, Roseburia, Ruminococcus, and Streptococcus, correlated with healthier eating behavior, while 5 genera, i.e., Clostridiales cluster IV and XIVb, Collinsella, Fusicatenibacter, and Parabacteroides, correlated with unhealthier eating (all | r | > 0.4, FDR-corrected p < 0.05). Some of these genera including Parabacteroides related to fiber intake and SCFA, and to weight status and treatment response in overweight/obese patients. In this exploratory analysis, specific bacterial genera, particularly Parabacteroides, were associated with weight status and eating behavior in two small, independent and well-characterized cross-sectional samples. These preliminary findings suggest two groups of presumably beneficial and unfavorable genera that relate to eating behavior and weight status, and indicate that dietary fiber and SCFA metabolism may modify these relationships. Larger interventional studies are needed to distinguish correlation from causation.
Collapse
|
132
|
Brown RM, Guerrero-Hreins E, Brown WA, le Roux CW, Sumithran P. Potential gut-brain mechanisms behind adverse mental health outcomes of bariatric surgery. Nat Rev Endocrinol 2021; 17:549-559. [PMID: 34262156 DOI: 10.1038/s41574-021-00520-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2021] [Indexed: 02/06/2023]
Abstract
Bariatric surgery induces sustained weight loss and metabolic benefits via notable effects on the gut-brain axis that lead to alterations in the neuroendocrine regulation of appetite and glycaemia. However, in a subset of patients, bariatric surgery is associated with adverse effects on mental health, including increased risk of suicide or self-harm as well as the emergence of depression and substance use disorders. The contributing factors behind these adverse effects are not well understood. Accumulating evidence indicates that there are important links between gut-derived hormones, microbial and bile acid profiles, and disorders of mood and substance use, which warrant further exploration in the context of changes in gut-brain signalling after bariatric surgery. Understanding the basis of these adverse effects is essential in order to optimize the health and well-being of people undergoing treatment for obesity.
Collapse
Affiliation(s)
- Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Eva Guerrero-Hreins
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Wendy A Brown
- Department of Surgery, Central Clinical School, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College, Dublin, Ireland
| | - Priya Sumithran
- Department of Medicine (St Vincent's), University of Melbourne, Melbourne, Victoria, Australia.
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia.
| |
Collapse
|
133
|
Rautmann AW, de La Serre CB. Microbiota's Role in Diet-Driven Alterations in Food Intake: Satiety, Energy Balance, and Reward. Nutrients 2021; 13:nu13093067. [PMID: 34578945 PMCID: PMC8470213 DOI: 10.3390/nu13093067] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota plays a key role in modulating host physiology and behavior, particularly feeding behavior and energy homeostasis. There is accumulating evidence demonstrating a role for gut microbiota in the etiology of obesity. In human and rodent studies, obesity and high-energy feeding are most consistently found to be associated with decreased bacterial diversity, changes in main phyla relative abundances and increased presence of pro-inflammatory products. Diet-associated alterations in microbiome composition are linked with weight gain, adiposity, and changes in ingestive behavior. There are multiple pathways through which the microbiome influences food intake. This review discusses these pathways, including peripheral mechanisms such as the regulation of gut satiety peptide release and alterations in leptin and cholecystokinin signaling along the vagus nerve, as well as central mechanisms, such as the modulation of hypothalamic neuroinflammation and alterations in reward signaling. Most research currently focuses on determining the role of the microbiome in the development of obesity and using microbiome manipulation to prevent diet-induced increase in food intake. More studies are necessary to determine whether microbiome manipulation after prolonged energy-dense diet exposure and obesity can reduce intake and promote meaningful weight loss.
Collapse
|
134
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
135
|
Hong J, Bo T, Xi L, Xu X, He N, Zhan Y, Li W, Liang P, Chen Y, Shi J, Li D, Yan F, Gu W, Wang W, Liu R, Wang J, Wang Z, Ning G. Reversal of Functional Brain Activity Related to Gut Microbiome and Hormones After VSG Surgery in Patients With Obesity. J Clin Endocrinol Metab 2021; 106:e3619-e3633. [PMID: 33950216 PMCID: PMC8372652 DOI: 10.1210/clinem/dgab297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 12/19/2022]
Abstract
CONTEXT Vertical sleeve gastrectomy (VSG) is becoming a prioritized surgical intervention for obese individuals; however, the brain circuits that mediate its effective control of food intake and predict surgical outcome remain largely unclear. OBJECTIVE We investigated VSG-correlated alterations of the gut-brain axis. METHODS In this observational cohort study, 80 patients with obesity were screened. A total of 36 patients together with 26 normal-weight subjects were enrolled and evaluated using the 21-item Three-Factor Eating Questionnaire (TFEQ), MRI scanning, plasma intestinal hormone analysis, and fecal sample sequencing. Thirty-two patients underwent VSG treatment and 19 subjects completed an average of 4-month follow-up evaluation. Data-driven regional homogeneity (ReHo) coupled with seed-based connectivity analysis were used to quantify VSG-related brain activity. Longitudinal alterations of body weight, eating behavior, brain activity, gastrointestinal hormones, and gut microbiota were detected and subjected to repeated measures correlation analysis. RESULTS VSG induced significant functional changes in the right putamen (PUT.R) and left supplementary motor area, both of which correlated with weight loss and TFEQ scores. Moreover, postprandial levels of active glucagon-like peptide-1 (aGLP-1) and Ghrelin were associated with ReHo of PUT.R; meanwhile, relative abundance of Clostridia increased by VSG was associated with improvements in aGLP-1 secretion, PUT.R activity, and weight loss. Importantly, VSG normalized excessive functional connectivities with PUT.R, among which baseline connectivity between PUT.R and right orbitofrontal cortex was related to postoperative weight loss. CONCLUSION VSG causes correlated alterations of gut-brain axis, including Clostridia, postprandial aGLP-1, PUT.R activity, and eating habits. Preoperative connectivity of PUT.R may represent a potential predictive marker of surgical outcome in patients with obesity.
Collapse
Affiliation(s)
- Jie Hong
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Tingting Bo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liuqing Xi
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | | | - Naying He
- Department of Radiology, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Yafeng Zhan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanyu Li
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Peiwen Liang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Yufei Chen
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Juan Shi
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Danjie Li
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Weiqiong Gu
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Weiqing Wang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Ruixin Liu
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Jiqiu Wang
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Zheng Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Guang Ning
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of Chinese Health Commission, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| |
Collapse
|
136
|
Jin ZL, Liu W. Progress in treatment of type 2 diabetes by bariatric surgery. World J Diabetes 2021; 12:1187-1199. [PMID: 34512886 PMCID: PMC8394224 DOI: 10.4239/wjd.v12.i8.1187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/29/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 2 diabetes (T2D) is increasing at an alarming rate worldwide. Bariatric surgical procedures, such as the vertical sleeve gastrectomy and Roux-en-Y gastric bypass, are the most efficient approaches to obtain substantial and durable remission of T2D. The benefits of bariatric surgery are realized through the consequent increased satiety and alterations in gastrointestinal hormones, bile acids, and the intestinal microbiota. A comprehensive understanding of the mechanisms by which various bariatric surgical procedures exert their benefits on T2D could contribute to the design of better non-surgical treatments for T2D. In this review, we describe the classification and evolution of bariatric surgery and explore the multiple mechanisms underlying the effect of bariatric surgery on insulin resistance. Based upon our summarization of the current knowledge on the underlying mechanisms, we speculate that the gut might act as a new target for improving T2D. Our ultimate goal with this review is to provide a better understanding of T2D pathophysiology in order to support development of T2D treatments that are less invasive and more scalable.
Collapse
Affiliation(s)
- Zhang-Liu Jin
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Liu
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
137
|
Ichikawa H, Imoto H, Tanaka N, Fujishima F, Tsuchiya T, Watanabe K, Aoki T, Kohyama A, Morikawa T, Ohnuma S, Naitoh T, Kamei T, Unno M. The role of bilio-pancreatic limb in nonalcoholic steatohepatitis improvement after duodenal-jejunal bypass in rats. Surgery 2021; 170:1006-1013. [PMID: 34389163 DOI: 10.1016/j.surg.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/14/2021] [Accepted: 07/02/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease, which is highly associated with obesity, includes nonalcoholic steatohepatitis. Lipopolysaccharides from the intestine would induce inflammation in the liver in nonalcoholic fatty liver disease. This study aimed to examine the role of the bilio-pancreatic limb in the effect of duodenal-jejunal bypass on nonalcoholic steatohepatitis, with respect to the gut-liver axis, using a rat model. METHODS Nonalcoholic steatohepatitis model rats were randomly assigned into 3 groups as follows: 1 sham group and 2 duodenal-jejunal bypass groups. The 2 duodenal-jejunal bypass groups were defined according to the bilio-pancreatic limb length: 30 cm (30-DJB group) and 0 cm (0-DJB group). Pathology findings and blood biochemistry, inflammatory cytokine and lipopolysaccharides receptor mRNA in the liver and common channel, and lipopolysaccharide-binding protein level in the portal vein were assessed. RESULTS The reduction in plasma alanine aminotransferase and nonalcoholic fatty liver disease activity score in the 30-DJB group was not observed in the 0-DJB group, similar to the sham group. In the liver tissue, mRNA of inflammatory cytokines and lipopolysaccharide receptors, the area occupied by CD68-positive macrophages, and the number of CD3-positive T-lymphocytes on immunostaining were lower in the 30-DJB group; however, these findings were not observed in the 0-DJB group, and lipopolysaccharide-binding protein levels in the portal vein and mRNA expressions of inflammation-related genes in the common channel showed similar tendencies. CONCLUSION The bilio-pancreatic limb plays an important role in the beneficial effect of duodenal-jejunal bypass for nonalcoholic steatohepatitis. The bilio-pancreatic limb may suppress lipopolysaccharides-related cascades in the liver by reducing intestinal inflammation.
Collapse
Affiliation(s)
- Hidetaka Ichikawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirofumi Imoto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Tanaka
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | - Takahiro Tsuchiya
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiro Watanabe
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Aoki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Kohyama
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Morikawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinobu Ohnuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Naitoh
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
138
|
Intestinal-derived FGF15 protects against deleterious effects of vertical sleeve gastrectomy in mice. Nat Commun 2021; 12:4768. [PMID: 34362888 PMCID: PMC8346483 DOI: 10.1038/s41467-021-24914-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Bariatric surgeries such as the Vertical Sleeve Gastrectomy (VSG) are invasive but provide the most effective improvements in obesity and Type 2 diabetes. We hypothesized a potential role for the gut hormone Fibroblast-Growth Factor 15/19 which is increased after VSG and pharmacologically can improve energy homeostasis and glucose handling. We generated intestinal-specific FGF15 knockout (FGF15INT-KO) mice which were maintained on high-fat diet. FGF15INT-KO mice lost more weight after VSG as a result of increased lean tissue loss. FGF15INT-KO mice also lost more bone density and bone marrow adipose tissue after VSG. The effect of VSG to improve glucose tolerance was also absent in FGF15INT-KO. VSG resulted in increased plasma bile acid levels but were considerably higher in VSG-FGF15INT-KO mice. These data point to an important role after VSG for intestinal FGF15 to protect the organism from deleterious effects of VSG potentially by limiting the increase in circulating bile acids. The mechanisms that mediate the effects of weight loss surgeries such as vertical sleeve gastrectomy (VSG) are incompletely understood. Here the authors show that intestinal FGF15 is necessary to improve glucose tolerance and to prevent the loss of muscle and bone mass after VSG, potentially via protection against bile acid toxicity.
Collapse
|
139
|
Toll-like receptor 4 and myeloid differentiation factor 88 are required for gastric bypass-induced metabolic effects. Surg Obes Relat Dis 2021; 17:1996-2006. [PMID: 34462225 PMCID: PMC9083208 DOI: 10.1016/j.soard.2021.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 01/04/2023]
Abstract
Background: Toll-like receptor 4 (TLR4) has been suggested as one of the forefront cross-communicators between the intestinal bacteria and the host to regulate inflammatory signals and energy homeostasis. High-fat diet–induced inflammation is mediated by changes in gut microbiota and requires a functional TLR-4, the deficiency of which renders mice resistant to diet-induced obesity and its associated metabolic dysfunction. Furthermore, gut microbiota was suggested to play a key role in the beneficial effects of Roux-en-Y gastric bypass (RYGB), a commonly performed bariatric procedure. Objectives: To explore whether TLR4, myeloid differentiation factor 8 (MyD88; 1 of its key downstream signaling regulators) and gut microbiota play an integrative role in RYGB-induced metabolic outcomes. Setting: Animal-based study. Method: We performed RYGB in TLR4 and MyD88 knock-out (KO) mice and used fecal microbiota transplant (FMT) from RYGB-operated animals to these genetic mouse models to address our questions. Results: We demonstrate that RYGB reduces TLR4 expression explicitly in the small and large intestine of C57Blc/6J mice. We also show that TLR4 KO mice have an attenuated glucoregulatory response to RYGB. In addition, we reveal that MyD88 KO mice fail to respond to all RYGB-induced metabolic effects. Finally, fecal microbiota transplant from RYGB-operated mice into TLR4 KO and MyD88 KO naïve recipients fails to induce a metabolic phenotype similar to that of the donors, as it does in wild-type recipients. Conclusion: TLR4 and MyD88 are required for RYGB-induced metabolic response that is likely mediated by gut microbiome.
Collapse
|
140
|
Karami R, Kermansaravi M, Pishgahroudsari M, Talebi M, Mohammadzadeh N, Pazouki A. Changes in gut microbial flora after Roux-en-Y gastric bypass and sleeve gastrectomy and their effects on post-operative weight loss. Updates Surg 2021; 73:1493-1499. [PMID: 33067675 DOI: 10.1007/s13304-020-00900-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Bariatric surgery affects gut microbial flora due to the anatomical and physiological changes it causes in the gastrointestinal tract. Understanding the interaction between the gut flora, the type of bariatric surgery and weight loss may help improve bariatric surgery outcomes. This study was designed to compare the effects of Roux-en-Y Gastric Bypass (RYGB) and Sleeve Gastrectomy (SG) on two main phyla of the gut microbiota in humans and evaluate their potential effect on weight changes. Thirty morbidly obese patients were divided into two groups and underwent laparoscopic SG or laparoscopic RYGB. The patients' weight changes and fecal samples were evaluated at baseline and 6 months after the surgery. A microbial flora count was carried out of the phyla Bacteroidetes and Firmicutes and Bacteroides Fragilis. Changes in the abundance of the flora and their correlation with weight loss were analyzed. After 6 months, the patients with a history of RYGB showed a significant decrease in stool Bacteroidetes while the reduction in the SG group was insignificant. Firmicutes abundance was almost unchanged following SG and RYGB. There was no significant change in Bacteroides Fragilis abundance in either of the two groups, but a positive correlation was observed between Bacteroides Fragilis and weight loss after SG and RYGB. Bariatric surgery can affect gut microbiota. It can be concluded that these changes are dependent on many factors and may play a role in weight loss.
Collapse
Affiliation(s)
- Reza Karami
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasool-E-Akram Hospital, Tehran, Iran
| | - Mohammad Kermansaravi
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasool-E-Akram Hospital, Tehran, Iran.
- Center of Excellence of of European Branch of International Federation for Surgery of Obesity, Hazrat-E-Rasool Hospital, Tehran, Iran.
| | - Mohaddese Pishgahroudsari
- BS, Statistics Analyzer, Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Mohammadzadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Pazouki
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasool-E-Akram Hospital, Tehran, Iran
- Center of Excellence of of European Branch of International Federation for Surgery of Obesity, Hazrat-E-Rasool Hospital, Tehran, Iran
| |
Collapse
|
141
|
Nayor M, Shah SH, Murthy V, Shah RV. Molecular Aspects of Lifestyle and Environmental Effects in Patients With Diabetes: JACC Focus Seminar. J Am Coll Cardiol 2021; 78:481-495. [PMID: 34325838 DOI: 10.1016/j.jacc.2021.02.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
Diabetes is characterized as an integrated condition of dysregulated metabolism across multiple tissues, with well-established consequences on the cardiovascular system. Recent advances in precision phenotyping in biofluids and tissues in large human observational and interventional studies have afforded a unique opportunity to translate seminal findings in models and cellular systems to patients at risk for diabetes and its complications. Specifically, techniques to assay metabolites, proteins, and transcripts, alongside more recent assessment of the gut microbiome, underscore the complexity of diabetes in patients, suggesting avenues for precision phenotyping of risk, response to intervention, and potentially novel therapies. In addition, the influence of external factors and inputs (eg, activity, diet, medical therapies) on each domain of molecular characterization has gained prominence toward better understanding their role in prevention. Here, the authors provide a broad overview of the role of several of these molecular domains in human translational investigation in diabetes.
Collapse
Affiliation(s)
- Matthew Nayor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/MattNayor
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/SvatiShah
| | - Venkatesh Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA. https://twitter.com/venkmurthy
| | - Ravi V Shah
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
142
|
Lee Y, Kim AH, Kim E, Lee S, Yu KS, Jang IJ, Chung JY, Cho JY. Changes in the gut microbiome influence the hypoglycemic effect of metformin through the altered metabolism of branched-chain and nonessential amino acids. Diabetes Res Clin Pract 2021; 178:108985. [PMID: 34329692 DOI: 10.1016/j.diabres.2021.108985] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 02/08/2023]
Abstract
AIMS Although metformin has been reported to affect the gut microbiome, the mechanism has not been fully determined. We explained the potential underlying mechanisms of metformin through a multiomics approach. METHODS An open-label and single-arm clinical trial involving 20 healthy Korean was conducted. Serum glucose and insulin concentrations were measured, and stool samples were collected to analyze the microbiome. Untargeted metabolomic profiling of plasma, urine, and stool samples was performed by GC-TOF-MS. Network analysis was applied to infer the mechanism of the hypoglycemic effect of metformin. RESULTS The relative abundances of Escherichia, Romboutsia, Intestinibacter, and Clostridium were changed by metformin treatment. Additionally, the relative abundances of metabolites, including carbohydrates, amino acids, and fatty acids, were changed. These changes were correlated with energy metabolism, gluconeogenesis, and branched-chain amino acid metabolism, which are major metabolic pathways related to the hypoglycemic effect. CONCLUSIONS We observed that specific changes in metabolites may affect hypoglycemic effects through both pathways related to AMPK activation and microbial changes. Energy metabolism was mainly related to hypoglycemic effects. In particular, branched-chain amino acid metabolism and gluconeogenesis were related to microbial metabolites. Our results will help uncover the potential underlying mechanisms of metformin through AMPK and the microbiome.
Collapse
Affiliation(s)
- Yujin Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea.
| | - Andrew HyoungJin Kim
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA.
| | - Eunwoo Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea.
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea.
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea.
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea; Clinical Trials Center, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.
| |
Collapse
|
143
|
Maksimaityte V, Bausys A, Kryzauskas M, Luksta M, Stundiene I, Bickaite K, Bausys B, Poskus T, Bausys R, Strupas K. Gastrectomy impact on the gut microbiome in patients with gastric cancer: A comprehensive review. World J Gastrointest Surg 2021; 13:678-688. [PMID: 34354801 PMCID: PMC8316847 DOI: 10.4240/wjgs.v13.i7.678] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/19/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common malignancies worldwide and gastrectomy remains the only potentially curative treatment option for this disease. However, the surgery leads to significant physiological and anatomical changes in the gastrointestinal (GI) tract including loss of the gastric barrier, an increase in oxygenation levels in the distal gut, and biliary diversion after gastrectomy. These changes in the GI tract influence the composition of the gut microbiome and thus, host health. Gastrectomy-induced dysbiosis is characterized by increased abundance of typical oral cavity bacteria, an increase in aero-tolerant bacteria (aerobes/facultative anaerobes), and increased abundance of bile acid-transforming bacteria. Furthermore, this dysbiosis is linked to intestinal inflammation, small intestinal bacterial overgrowth, various GI symptoms, and an increased risk of colorectal cancer.
Collapse
Affiliation(s)
- Vaidota Maksimaityte
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | - Augustinas Bausys
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius 08660, Lithuania
| | - Marius Kryzauskas
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | - Martynas Luksta
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | - Ieva Stundiene
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | | | - Bernardas Bausys
- Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | - Tomas Poskus
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| | - Rimantas Bausys
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius 08660, Lithuania
| | - Kestutis Strupas
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius 03101, Lithuania
| |
Collapse
|
144
|
Juárez-Fernández M, Román-Sagüillo S, Porras D, García-Mediavilla MV, Linares P, Ballesteros-Pomar MD, Urioste-Fondo A, Álvarez-Cuenllas B, González-Gallego J, Sánchez-Campos S, Jorquera F, Nistal E. Long-Term Effects of Bariatric Surgery on Gut Microbiota Composition and Faecal Metabolome Related to Obesity Remission. Nutrients 2021; 13:nu13082519. [PMID: 34444679 PMCID: PMC8397959 DOI: 10.3390/nu13082519] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Obesity is one of the main worldwide public health concerns whose clinical management demands new therapeutic approaches. Bariatric surgery is the most efficient treatment when other therapies have previously failed. Due to the role of gut microbiota in obesity development, the knowledge of the link between bariatric surgery and gut microbiota could elucidate new mechanistic approaches. This study aims to evaluate the long-term effects of bariatric surgery in the faecal metagenome and metabolome of patients with severe obesity. Faecal and blood samples were collected before and four years after the intervention from patients with severe obesity. Biochemical, metagenomic and metabolomic analyses were performed and faecal short-chain fatty acids were measured. Bariatric surgery improved the obesity-related status of patients and significantly reshaped gut microbiota composition. Moreover, this procedure was associated with a specific metabolome profile characterized by a reduction in energetic and amino acid metabolism. Acetate, butyrate and propionate showed a significant reduction with bariatric surgery. Finally, correlation analysis suggested the existence of a long-term compositional and functional gut microbiota profile associated with the intervention. In conclusion, bariatric surgery triggered long-lasting effects on gut microbiota composition and faecal metabolome that could be associated with the remission of obesity.
Collapse
Affiliation(s)
- María Juárez-Fernández
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - Sara Román-Sagüillo
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - David Porras
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
| | - María Victoria García-Mediavilla
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pedro Linares
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - María Dolores Ballesteros-Pomar
- Departamento de Endocrinología y Nutrición, Complejo Asistencial Universitario de León, 24071 León, Spain; (M.D.B.-P.); (A.U.-F.)
| | - Ana Urioste-Fondo
- Departamento de Endocrinología y Nutrición, Complejo Asistencial Universitario de León, 24071 León, Spain; (M.D.B.-P.); (A.U.-F.)
| | - Begoña Álvarez-Cuenllas
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - Javier González-Gallego
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sonia Sánchez-Campos
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Jorquera
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Aparato Digestivo, Complejo Asistencial Universitario de León, 24071 León, Spain; (P.L.); (B.Á.-C.)
| | - Esther Nistal
- Instituto Universitario de Biomedicina (IBIOMED), Universidad de León, 24007 León, Spain; (M.J.-F.); (S.R.-S.); (D.P.); (M.V.G.-M.); (J.G.-G.); (S.S.-C.); (F.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-9-8729-1997
| |
Collapse
|
145
|
Dong TS, Luu K, Lagishetty V, Sedighian F, Woo SL, Dreskin BW, Katzka W, Chang C, Zhou Y, Arias-Jayo N, Yang J, Ahdoot AI, Ye J, Li Z, Pisegna JR, Jacobs JP. The Intestinal Microbiome Predicts Weight Loss on a Calorie-Restricted Diet and Is Associated With Improved Hepatic Steatosis. Front Nutr 2021; 8:718661. [PMID: 34307440 PMCID: PMC8295485 DOI: 10.3389/fnut.2021.718661] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The microbiome has been shown in pre-clinical and epidemiological studies to be important in both the development and treatment of obesity and metabolic associated fatty liver disease (MAFLD). However, few studies have examined the role of the microbiome in the clinical response to calorie restriction. To explore this area, we performed a prospective study examining the association of the intestinal microbiome with weight loss and change in hepatic steatosis on a calorie-restricted diet. Methods: A prospective dietary intervention study of 80 overweight and obese participants was performed at the Greater West Los Angeles Veterans Affair Hospital. Patients were placed on a macronutrient standardized diet for 16 weeks, including 14 weeks of calorie restriction (500 calorie deficit). Body composition analysis by impedance, plasma lipid measurements, and ultrasound elastography to measure hepatic steatosis were performed at baseline and week 16. Intestinal microbiome composition was assessed using 16S rRNA gene sequencing. A per protocol analysis was performed on all subjects completing the trial (n = 46). Results: Study completers showed significant reduction in weight, body mass index, total cholesterol, low density lipoprotein, and triglyceride. Subjects who lost at least 5% of their body weight had significantly greater reduction in serum triglyceride and hepatic steatosis than those with <5% body weight loss. Enterococcus and Klebsiella were reduced at the end of the trial while Coprococcus and Collinsella were increased. There were also significant baseline microbiome differences between patients who had at least 5% weight loss as compared to those that did not. Lachnoclostridium was positively associated with hepatic steatosis and Actinomyces was positively associated with hepatic steatosis and weight. Baseline microbiome profiles were able to predict which patients lost at least 5% of their body weight with an AUROC of 0.80. Conclusion: Calorie restriction alters the intestinal microbiome and improves hepatic steatosis in those who experience significant weight loss. Baseline microbiome differences predict weight loss on a calorie-restricted diet and are associated with improvement in hepatic steatosis, suggesting a role of the gut microbiome in mediating the clinical response to calorie restriction.
Collapse
Affiliation(s)
- Tien S. Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Kayti Luu
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Farzaneh Sedighian
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Shih-Lung Woo
- Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Benjamin W. Dreskin
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - William Katzka
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Candace Chang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yi Zhou
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nerea Arias-Jayo
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julianne Yang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aaron I. Ahdoot
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jason Ye
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhaoping Li
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Center for Human Nutrition, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joseph R. Pisegna
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Jonathan P. Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
146
|
Cunningham AL, Stephens JW, Harris DA. A review on gut microbiota: a central factor in the pathophysiology of obesity. Lipids Health Dis 2021; 20:65. [PMID: 34233682 PMCID: PMC8262044 DOI: 10.1186/s12944-021-01491-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications constitute a substantial burden. Considerable published research describes the novel relationships between obesity and gut microbiota communities. It is becoming evident that microbiota behave in a pivotal role in their ability to influence homeostatic mechanisms either to the benefit or detriment of host health, the extent of which is not fully understood. A greater understanding of the contribution of gut microbiota towards host pathophysiology is revealing new therapeutic avenues to tackle the global obesity epidemic. This review focuses on causal relationships and associations with obesity, proposed central mechanisms encouraging the development of obesity and promising prospective methods for microbiota manipulation.
Collapse
Affiliation(s)
- A L Cunningham
- Department of Surgery, Swansea Bay University Health Board, Swansea, SA2 8QA, UK. .,Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK.
| | - J W Stephens
- Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK
| | - D A Harris
- Department of Surgery, Swansea Bay University Health Board, Swansea, SA2 8QA, UK.,Swansea University Medical School, Swansea University, Swansea, SA2 8QA, UK
| |
Collapse
|
147
|
Li X, Yang L, Li J, Lin L, Zheng G. A flavonoid-rich Smilax china L. extract prevents obesity by upregulating the adiponectin-receptor/AMPK signalling pathway and modulating the gut microbiota in mice. Food Funct 2021; 12:5862-5875. [PMID: 34019043 DOI: 10.1039/d1fo00282a] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The aim of this study was to investigate the effects of Smilax china L. flavonoid (SCF) on obesity and changes in gut microbiota high-fat/high-sucrose (HFHS)-fed mice. Male C57BL/6 mice fed either a normal-chow (NC) or a HFHS diet were treated with SCF for 12 weeks. The effect of SCF on the composition of gut microbiota was assessed by 16S rDNA sequencing. SCFA levels in the caecum were quantified by GC-MS. SCF supplementation alleviated the body weight gain, fat accumulation, serum lipid parameters, and hepatic steatosis and improved glucose homeostasis. SCF significantly increased plasma adiponectin level, adiponectin-receptor-gene (AdipoR1 and AdipoR2) expression in the liver, activated AMPKα, downregulated the expression of SREBP1-c, FAS, and ACCα, and upregulated the expression of PPARα, CPT-1α, and UCP-1. The anti-obesity effects of SCF might be through upregulation of adiponectin-receptor/AMPK signalling to improve lipid metabolism. SCF reversed HFHS-induced dysbiosis of gut microbiota and decreased SCFA production in the caecum, thus reducing energy absorption and leading to loss of body weight. Spearman's correlation analysis revealed significant correlations between obesity phenotypes, SCFA levels, and changes in gut microbiota. The results showed that SCF may be an effective dietary supplement that is useful for suppressing the development of obesity and associated disorders.
Collapse
Affiliation(s)
- Xin Li
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Licong Yang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Jingen Li
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Lezhen Lin
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
148
|
Miller MO, Kashyap PC, Becker SL, Thomas RM, Hodin RA, Miller G, Hundeyin M, Pushalkar S, Cohen D, Saxena D, Shogan BD, Morris-Stiff GJ. SSAT State-of-the-Art Conference: Advancements in the Microbiome. J Gastrointest Surg 2021; 25:1885-1895. [PMID: 32989690 DOI: 10.1007/s11605-020-04551-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The microbiome plays a major role in human physiology by influencing obesity, inducing inflammation, and impacting cancer therapies. During the 60th Annual Meeting of the Society of the Alimentary Tract (SSAT) at the State-of-the-Art Conference, experts in the field discussed the influence of the microbiome. This paper is a summary of the influence of the microbiome on obesity, inflammatory bowel disease, pancreatic cancer, cancer therapies, and gastrointestinal optimization. This review shows how the microbiome plays an important role in the development of diseases and surgical complications. Future studies are needed in targeting the gut microbiome to develop individualized therapies.
Collapse
Affiliation(s)
- Miquell O Miller
- Department of General Surgery, Stanford University, 300 Pasteur Dr, Stanford, CA, 94305, USA.
| | - Purna C Kashyap
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sarah L Becker
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ryan M Thomas
- Departments of Surgery, Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Richard A Hodin
- Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - George Miller
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Mautin Hundeyin
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Smruti Pushalkar
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Deirdre Cohen
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Benjamin D Shogan
- Department of Surgery, University of Chicago, Chicago, IL, 60637, USA
| | | |
Collapse
|
149
|
Barbu E, Popescu MR, Popescu AC, Balanescu SM. Phenotyping the Prediabetic Population-A Closer Look at Intermediate Glucose Status and Cardiovascular Disease. Int J Mol Sci 2021; 22:6864. [PMID: 34202289 PMCID: PMC8268766 DOI: 10.3390/ijms22136864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
Even though the new thresholds for defining prediabetes have been around for more than ten years, there is still controversy surrounding the precise characterization of this intermediate glucose metabolism status. The risk of developing diabetes and macro and microvascular disease linked to prediabetes is well known. Still, the prediabetic population is far from being homogenous, and phenotyping it into less heterogeneous groups might prove useful for long-term risk assessment, follow-up, and primary prevention. Unfortunately, the current definition of prediabetes is quite rigid and disregards the underlying pathophysiologic mechanisms and their potential metabolic progression towards overt disease. In addition, prediabetes is commonly associated with a cluster of risk factors that worsen the prognosis. These risk factors all revolve around a common denominator: inflammation. This review focuses on identifying the population that needs to be screened for prediabetes and the already declared prediabetic patients who are at a higher risk of cardiovascular disease and require closer monitoring.
Collapse
Affiliation(s)
| | - Mihaela-Roxana Popescu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 011461 Bucharest, Romania; (E.B.); (S.-M.B.)
| | - Andreea-Catarina Popescu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 011461 Bucharest, Romania; (E.B.); (S.-M.B.)
| | | |
Collapse
|
150
|
Signorini FJ, Arocena L, Viscido G, Obeide L, Gorodner V, Moser F. Re-emergence of Diabetes After Sleeve Gastrectomy in Patients with Long-Term Follow-up. Indian J Surg 2021. [DOI: 10.1007/s12262-021-02987-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|